ATL8 typically refers to a membrane-localized ubiquitin ligase in plants, involved in sugar signaling and stress responses . No antibodies targeting this protein are explicitly documented in the provided sources.
RS-ATL8 is a humanized rat basophilic leukemia reporter cell line used to detect allergen-specific IgE cross-linking in shellfish allergy studies . This tool is unrelated to ATL8 antibodies but shares a similar nomenclature.
ACTL8 (a distinct gene/protein) has commercial antibodies, such as Abcam’s Anti-ACTL8 antibody (ab184562) , but this is unrelated to ATL8.
The RS-ATL8 system is a humanized basophilic leukemia model used to identify allergens capable of IgE cross-linking. Key findings from shrimp allergy studies include:
While no ATL8-specific antibodies are documented, insights from related systems highlight antibody development strategies:
A well-characterized monoclonal antibody used in Alzheimer’s disease research:
A commercial antibody for the unrelated ACTL8 protein:
| Application | Details |
|---|---|
| Detection | Western blot (1/2000 dilution) in human testis lysate . |
| Predicted Band | 41 kDa (consistent with ACTL8’s molecular weight) . |
ATL8 Antibody Development:
No antibodies targeting ATL8 are reported in literature or commercial catalogs.
Potential strategies: Recombinant ATL8 production, epitope mapping, and hybridoma technology.
Cross-Disciplinary Applications:
Leveraging RS-ATL8’s IgE detection system for allergen characterization in food or environmental contexts.
Exploring ATL8’s role in plant biology (e.g., stress responses) using molecular probes.
ATL8 is a plant-specific RING-type ubiquitin ligase belonging to the Arabidopsis Tóxicos en Levadura (ATL) family, which currently comprises 91 identified isoforms in the Arabidopsis genome . This membrane-localized protein plays a crucial role in plants' adaptation to sugar starvation stress, with its expression significantly increasing under extended darkness or sugar deprivation conditions and decreasing rapidly when exogenous sugar is supplied .
Antibodies against ATL8 are essential tools that enable researchers to:
Track protein abundance during stress responses
Investigate subcellular localization through immunohistochemistry
Identify protein interaction partners through co-immunoprecipitation
Study the regulatory mechanisms governing protein degradation pathways
Recent research has identified Starch Synthase 4 as a putative interaction partner of ATL8, suggesting its involvement in regulating starch accumulation based on sugar availability . Antibodies provide a direct means of validating such interactions and exploring their functional significance in plant physiology.
Understanding ATL8's structural features is critical for developing effective antibodies and experimental approaches. ATL8 contains:
A single transmembrane-like hydrophobic amino acid region (residues 31-53) at its N-terminus
A RING-H2 type zinc finger domain in its middle portion
A conserved cysteine residue (C123) essential for ubiquitin ligase activity
These structural characteristics create specific challenges and opportunities for antibody production and application:
The N-terminal hydrophobic region presents difficulties for recombinant protein expression, often necessitating its exclusion when producing immunogens .
The highly conserved RING domain may lead to cross-reactivity with other ATL family members, requiring careful epitope selection to ensure specificity.
For functional studies, antibodies targeting the RING domain may interfere with ubiquitin ligase activity, which can be either a limitation or a useful tool depending on the experimental goals.
When designing constructs for recombinant protein production as immunogens, researchers have successfully used ATL8 fragments beginning from residue 71 (valine) to ensure proper expression .
For antibody validation, the catalytically inactive ATL8C123S mutant provides an excellent control to distinguish between functional and structural recognition .
Proper validation of ATL8 antibodies requires rigorous controls to ensure specificity, particularly given the existence of 91 ATL family members in Arabidopsis with potential structural similarities . Essential validation steps include:
Genetic controls:
Biochemical controls:
Technical controls:
Analytical validation:
Researchers investigating ATL8 expression during sugar starvation can employ several complementary techniques:
RT-PCR analysis:
Western blot analysis:
Requires careful membrane protein extraction protocols
Benefits from comparison between wild-type and mutant plants
Should include appropriate controls for equal loading
Can demonstrate protein-level regulation that may differ from transcript patterns
Immunolocalization:
Enables visualization of subcellular distribution in membrane-bound compartments
Can be combined with markers for specific organelles to determine precise localization
Allows detection of potential relocalization under stress conditions
Requires careful fixation protocols to preserve membrane protein epitopes
Fluorescent protein fusion approaches:
These methods have revealed that ATL8 expression is primarily regulated by cellular sugar availability rather than light signaling pathways, as demonstrated by the absence of increased expression during extended darkness when exogenous sucrose is present .
Studying the ubiquitin ligase activity of ATL8 requires specific methodological approaches:
Recombinant protein production:
In vitro ubiquitination assay setup:
Include purified E1 (ubiquitin-activating enzyme)
Add appropriate E2 (ubiquitin-conjugating enzyme)
Provide ubiquitin and ATP
Incubate at physiological temperature (typically 30°C for plant proteins)
Detection and analysis:
Use Western blotting with anti-ubiquitin antibodies to detect ubiquitinated products
Look for heterogeneous higher molecular weight bands indicating ubiquitination
Include time course analysis (0, 30, 120 minutes) to monitor reaction progression
Compare wild-type ATL8 activity with the catalytically inactive ATL8C123S mutant
Substrate-specific assays:
This approach has successfully demonstrated that ATL8 possesses RING-type ubiquitin ligase activity in vitro, which is abolished when the conserved cysteine residue (C123) is mutated to serine .
Identifying and validating protein interactions with ATL8 requires multiple complementary approaches:
Co-immunoprecipitation with mass spectrometry:
Targeted co-immunoprecipitation:
Use antibodies against ATL8 to pull down potential interaction partners
Alternatively, use antibodies against suspected partners to co-precipitate ATL8
Confirm interactions by Western blotting
Include appropriate negative controls (pre-immune serum, unrelated antibodies)
Bimolecular Fluorescence Complementation (BiFC):
Fuse ATL8 to one half of a split fluorescent protein
Fuse candidate interactors to the complementary half
Co-express in plant cells and observe for reconstituted fluorescence
Include appropriate controls with non-interacting proteins
Yeast two-hybrid assays:
Use membrane-based yeast two-hybrid systems suitable for membrane proteins
Create constructs lacking the transmembrane domain for conventional Y2H
Screen against cDNA libraries or test specific candidate interactions
Validate positive interactions in planta using methods above
Protein modification analysis:
Determine if ATL8 ubiquitinates identified interactors
Monitor stability of interacting proteins in wild-type vs. ATL8 mutant plants
Assess whether interactions are regulated by sugar availability
Investigate co-localization during stress conditions
These approaches should be applied iteratively, with initial identification of candidates followed by validation through multiple independent methods.
ATL8 antibodies enable sophisticated investigations into how sugar starvation triggers selective protein degradation:
Temporal analysis of protein degradation:
Track ATL8 protein levels alongside potential substrates during sugar starvation
Monitor ubiquitination status of target proteins using anti-ubiquitin antibodies
Measure protein half-lives in wild-type versus ATL8 mutant plants
Create time-course profiles under different sugar deprivation regimes
Regulatory pathway investigation:
Use phospho-specific antibodies to monitor SnRK1 activation alongside ATL8 expression
SnRK1 knockdown mutants show reduced ATL8 expression, suggesting this energy sensor acts upstream of ATL8
Investigate how ATL8 interacts with other components of energy sensing pathways
Examine relationships with autophagy and proteasomal degradation systems
Organelle-specific degradation analysis:
Use subcellular fractionation followed by immunoblotting
Compare degradation patterns in different cellular compartments
Study co-localization of ATL8 with substrate proteins during starvation
Investigate whether ATL8 mediates organelle-specific protein quality control
Metabolic impact assessment:
Correlate ATL8 expression with changes in starch metabolism
Monitor the relationship with branched-chain amino acid (BCAA) catabolism enzymes
ATL8 expression is highly coordinated with genes involved in BCAA degradation (IVD, BCE2/DIN3, DIN4, MCCB)
Investigate how these coordinated pathways contribute to maintaining electron flow to the respiratory chain during sugar limitation
The identification of Starch Synthase 4 (SS4) as a putative ATL8 interactor suggests a direct role in modulating starch accumulation in response to sugar availability . A comprehensive experimental design to investigate this relationship would include:
Protein interaction verification:
Confirm ATL8-SS4 interaction using reciprocal co-immunoprecipitation
Determine domains responsible for interaction using truncation mutants
Investigate whether interaction is regulated by sugar availability
Use fluorescence resonance energy transfer (FRET) to quantify interaction dynamics
Functional relationship characterization:
Generate single and double mutants of ATL8 and SS4
Measure starch content under normal and sugar starvation conditions
Analyze SS4 protein levels and stability in wild-type versus ATL8 mutant plants
Determine if ATL8 ubiquitinates SS4 in vitro and in vivo
Temporal and spatial analysis:
Monitor co-localization of ATL8 and SS4 during light/dark transitions
Track protein dynamics during extended darkness using time-course immunoblotting
Investigate tissue-specific interactions in different plant organs
Analyze chloroplast morphology and starch granule formation
Physiological relevance assessment:
Measure survival rates during extended darkness in wild-type versus mutant plants
Analyze carbon allocation and utilization patterns during energy stress
Investigate recovery dynamics when sugar becomes available after starvation
Examine starch degradation rates and patterns
Integration with energy sensing pathways:
Determine how SnRK1 signaling affects the ATL8-SS4 interaction
Investigate coordination with trehalose-6-phosphate signaling
Analyze transcriptional coordination with other starch metabolism genes
Study potential feedback mechanisms regulating ATL8 expression based on starch status
ATL8 antibodies provide valuable tools for exploring how sugar starvation response integrates with other stress signaling networks:
Combined stress experiments:
Subject plants to sugar starvation plus additional stresses (cold, drought, salt)
Use immunoblotting to monitor ATL8 protein levels under combined stresses
Compare with related ATL family members like ATL80 (involved in phosphate mobilization and cold stress)
Investigate stress-specific post-translational modifications
Hormone response integration:
Analyze how plant hormones affect ATL8 expression and localization
Study ATL8 regulation in hormone signaling mutants
Explore co-immunoprecipitation to identify hormone-dependent interaction partners
Investigate whether hormone treatments alter ATL8's ubiquitin ligase activity
Signaling pathway cross-regulation:
Use phospho-specific antibodies to monitor energy sensors (SnRK1) alongside stress-activated protein kinases
Analyze whether stress-responsive transcription factors bind to the ATL8 promoter
Study how alternative stress-induced membrane modifications affect ATL8 localization
Determine if ATL8 substrates change under different stress combinations
Multi-omics integration:
Correlate ATL8 protein levels with transcriptome changes during combined stresses
Analyze the ubiquitinome in wild-type versus ATL8 mutant plants under different stresses
Study metabolome adjustments focusing on carbon allocation and amino acid metabolism
Develop network models of ATL8's position within integrated stress response systems
This approach reveals how ATL8 functions within the broader context of plant stress adaptation mechanisms, potentially identifying convergence points where multiple stress responses are coordinated through targeted protein degradation.
Researchers working with ATL8 antibodies frequently encounter several technical challenges:
Low signal intensity in Western blots:
High background in immunolocalization:
Problem: Non-specific binding to membranes
Solution: Optimize blocking conditions (BSA vs. milk, concentration, duration)
Problem: Plant tissue autofluorescence
Solution: Use appropriate filters and fluorophores with emission spectra distinct from autofluorescence
Cross-reactivity with other ATL family members:
Problem: Antibodies detecting related ATL proteins
Solution: Pre-absorb antibodies with recombinant proteins of closely related ATLs
Problem: Difficulty distinguishing specific signal
Solution: Include ATL8 knockout/knockdown plants as negative controls
Inconsistent immunoprecipitation results:
Variable results between experiments:
Problem: Antibody batch-to-batch variation
Solution: Validate each new antibody lot against standard samples
Problem: Environmental factors affecting ATL8 expression
Solution: Strictly control growth conditions and sugar availability
Optimizing immunohistochemistry for membrane-localized proteins like ATL8 requires specific protocol adjustments:
Fixation optimization:
Test multiple fixatives (paraformaldehyde, glutaraldehyde, combinations)
Evaluate different fixation durations (30 minutes to overnight)
Consider mild fixation to preserve membrane protein epitopes
Test with and without vacuum infiltration for better fixative penetration
Membrane permeabilization:
Use detergents appropriate for membrane proteins (Triton X-100, Tween-20, saponin)
Test concentration gradients to determine optimal permeabilization
Consider detergent-free methods using freeze-thaw cycles for certain applications
Evaluate enzymatic digestion of cell walls for improved antibody accessibility
Antigen retrieval methods:
Test heat-induced epitope retrieval at various pH conditions
Evaluate proteolytic retrieval methods (proteinase K, trypsin)
Consider sodium citrate buffer treatment
Compare microwave, pressure cooker, and water bath methods
Signal amplification strategies:
Use biotin-streptavidin systems for signal enhancement
Consider tyramide signal amplification for low-abundance proteins
Test polymer-based detection systems
Evaluate quantum dot conjugates for improved signal-to-noise ratio
Controls and validation:
Include wild-type versus ATL8 mutant tissue sections
Compare with GFP-tagged ATL8 localization patterns
Perform peptide competition controls
Include secondary-only controls to assess non-specific binding
These optimizations help ensure specific detection of membrane-localized ATL8 while minimizing background and preserving tissue morphology.
The dynamic and often transient nature of ubiquitin ligase-substrate interactions presents challenges for studying ATL8's biological targets:
Stabilization of interactions:
Use catalytically inactive ATL8C123S mutant which forms more stable complexes with substrates
Apply reversible crosslinking before cell lysis (formaldehyde, DSP, DTBP)
Treat samples with proteasome inhibitors to prevent substrate degradation
Consider 26S proteasome mutants to stabilize ubiquitinated intermediates
Proximity-based labeling approaches:
Fuse ATL8 to biotin ligase (BioID) or engineered peroxidase (APEX)
Allow in vivo biotinylation of proximal proteins
Purify biotinylated proteins and identify by mass spectrometry
Compare labeling patterns between wild-type ATL8 and ATL8C123S
Synchronized induction systems:
Create inducible ATL8 expression systems
Apply sugar starvation conditions in a controlled time course
Capture early interaction events before substrate degradation
Perform time-resolved interaction proteomics
Computational prediction and targeted validation:
Use bioinformatics to predict potential ATL8 substrates based on degron motifs
Screen candidates systematically using co-immunoprecipitation
Validate with in vitro ubiquitination assays
Confirm physiological relevance through genetic approaches
Specialized co-immunoprecipitation protocols:
Use tandem affinity purification with stringent washing
Include detergents optimized for membrane protein interactions
Apply TUBE (Tandem Ubiquitin Binding Entities) technology to capture ubiquitinated substrates
Consider on-bead digestion followed by mass spectrometry to minimize sample manipulation
These approaches help overcome the inherent challenges of studying enzyme-substrate interactions that are both transient and often lead to substrate degradation.
Quantification methodology:
Use densitometry software with appropriate background subtraction
Measure integrated density rather than peak intensity
Establish standard curves using recombinant protein dilutions
Normalize to appropriate loading controls (total protein stains preferred over housekeeping proteins)
Experimental design considerations:
Include at least three biological replicates per condition
Analyze technical replicates to assess method variability
Use randomized blot loading patterns to avoid edge effects
Include positive and negative controls on each blot
Statistical tests for comparative studies:
Apply ANOVA for comparing multiple conditions
Use appropriate post-hoc tests (Tukey's, Bonferroni, Dunnett's)
Consider non-parametric alternatives if normality assumptions are violated
Perform power analysis to ensure adequate sample size
Time-course analysis approaches:
Apply repeated measures ANOVA for within-subject designs
Consider curve-fitting for expression kinetics
Use area under curve (AUC) calculations for cumulative response
Apply time series analysis for complex temporal patterns
| Statistical Approach | Application | Advantages | Limitations |
|---|---|---|---|
| Student's t-test | Comparing two conditions | Simple, widely understood | Only for two groups, assumes normality |
| One-way ANOVA | Multiple condition comparison | Accounts for multiple testing | Requires post-hoc testing |
| Two-way ANOVA | Testing effects of two factors | Detects interaction effects | More complex interpretation |
| Repeated measures ANOVA | Time course studies | Accounts for within-subject correlation | Requires sphericity |
| Non-parametric tests | Non-normal distributions | No normality assumption | Less statistical power |
| Regression analysis | Continuous relationships | Models response curves | Assumes linearity (unless specified) |
Distinguishing genuine ATL8 signals from artifacts in immunohistochemistry requires systematic validation and controls:
Essential controls for signal validation:
Genetic controls: Compare wild-type with ATL8 knockout/knockdown tissues
Antibody controls: Pre-immune serum, isotype controls, secondary-only controls
Peptide competition: Pre-absorb antibody with immunizing peptide
Expression controls: Compare tissues with known high versus low ATL8 expression
Common artifacts and their resolution:
Autofluorescence: Identify through multi-channel imaging and spectral unmixing
Edge effects: Examine pattern distribution relative to tissue architecture
Fixation artifacts: Compare multiple fixation protocols
Non-specific binding: Optimize blocking conditions systematically
Validation through complementary approaches:
Confirm patterns with ATL8-GFP fusion protein localization
Verify with subcellular fractionation followed by immunoblotting
Support with in situ hybridization for transcript localization
Compare with published expression data from similar conditions
Quantitative assessment approaches:
Use digital image analysis with defined intensity thresholds
Perform colocalization analysis with known compartment markers
Apply unbiased stereological methods for pattern quantification
Implement machine learning algorithms for pattern recognition
These systematic approaches ensure that immunohistochemical findings represent genuine ATL8 localization rather than technical artifacts or non-specific signals.
Integrating ATL8 protein-level data with transcriptomic information provides deeper insights into regulatory mechanisms:
Correlation analysis approaches:
Compare ATL8 protein levels with mRNA expression under identical conditions
Identify potential post-transcriptional regulation when protein and mRNA levels diverge
Correlate with expression patterns of genes involved in BCAA catabolism that show coordinated regulation
Analyze time lags between transcriptional and protein-level changes
Regulatory network reconstruction:
Identify transcription factors potentially controlling both ATL8 and co-regulated genes
Analyze promoter elements of coordinately regulated genes
Study the effects of SnRK1 signaling on both transcriptome and ATL8 protein levels
Map potential feedback mechanisms where protein degradation influences transcription
Pathway enrichment integration:
Perform gene ontology analysis on genes co-regulated with ATL8
Identify biological processes enriched in both transcriptomic and proteomic datasets
Apply pathway analysis to position ATL8 within stress response networks
Use protein interaction data to connect transcriptional modules
Visualization and modeling approaches:
Develop integrated network visualizations combining protein and transcript data
Create mathematical models predicting ATL8 dynamics based on multiple data types
Apply machine learning to identify patterns across multi-omics datasets
Implement Bayesian network analysis to infer causal relationships
This integration provides a systems-level understanding of how ATL8 functions within the broader context of plant stress response pathways, revealing both transcriptional and post-translational regulatory mechanisms.